Synthesis and Pharmacological Evaluation of 3-(3, 4-Dichlorophenyl

Apr 14, 2004 - Christina M. Dersch,| Judith L. Flippen-Anderson,#,⊥ Damon Parrish,# Arthur E. Jacobson,‡ and. Kenner C. Rice*,‡. Laboratory of M...
0 downloads 0 Views 234KB Size
2624

J. Med. Chem. 2004, 47, 2624-2634

Synthesis and Pharmacological Evaluation of 3-(3,4-Dichlorophenyl)-1-indanamine Derivatives as Nonselective Ligands for Biogenic Amine Transporters Han Yu,‡,§ In Jong Kim,‡ John E. Folk,‡ Xinrong Tian,‡,† Richard B. Rothman,| Michael H. Baumann,| Christina M. Dersch,| Judith L. Flippen-Anderson,#,⊥ Damon Parrish,# Arthur E. Jacobson,‡ and Kenner C. Rice*,‡ Laboratory of Medicinal Chemistry, Building 8, Room B1-23, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Department of Health and Human Services, Bethesda, Maryland, 20892-0815, Clinical Psychopharmacology Section, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, Maryland 21224, and Laboratory for the Structure of Matter, Naval Research Laboratory, Washington, D.C. 20375 Received November 21, 2003

In our efforts toward developing a nonselective ligand that would block the effects of stimulants such as methamphetamine at dopamine (DA), serotonin (5-HT), and norepinephrine (NE) transporters, we synthesized a series of 3-(3,4-dichlorophenyl)-1-indanamine derivatives. Two of the examined higher affinity compounds had a phenolic hydroxyl group enabling preparation of a medium to long chain carboxylic acid ester that might eventually be useful for a longacting depot formulation. The in vitro data indicated that (-)-(1R,3S)-trans-3-(3,4-dichlorophenyl)-6-hydroxy-N-methyl-1-indanamine ((-)-(1R,3S)-11) displays high-affinity binding and potent inhibition of uptake at all three biogenic amine transporters. In vivo microdialysis experiments demonstrated that intravenous administration of (-)-(1R,3S)-11 to rats elevated extracellular DA and 5-HT in the nucleus accumbens in a dose-dependent manner. Pretreating rats with 0.5 mg/kg (-)-(1R,3S)-11 elevated extracellular DA and 5-HT by approximately 150% and reduced methamphetamine-induced neurotransmitter release by about 50%. Ex vivo autoradiography, however, demonstrated that iv administration of (-)-(1R,3S)-11 produced a dose-dependent, persistent occupation of 5-HT transporter binding sites but not DA transporter sites. Introduction The abuse of methamphetamine and other amphetamine-like stimulants has had a disturbing effect on public health in the United States.1-3 Studies have shown that methamphetamine can act as a substrate for transporters of the biogenic amine neurotransmitters, dopamine (DA), serotonin (5-HT), and norepinephrine (NE), and can be transported by these transmembrane proteins into the nerve terminal.4 The substrate activity of methamphetamine and related agents evokes the nonexocytotic release of biogenic amines transmitters. Increased release of DA in the central nervous system is thought to mediate the dependence-producing effects of methamphetamine, whereas increased release of NE in both the peripheral and central nervous system is thought to induce undesirable cardiovascular side effects.5 However, recent data suggest that norepinephrine may play an important role in mediating amphet* To whom correspondence should be addressed. Phone: (301) 496 1856. Fax: (301) 402 0589. E-mail: [email protected]. ‡ National Institute of Diabetes and Digestive and Kidney Disease. † Present address: Procter & Gamble Pharmaceuticals, HCRC, Mason, OH 45040. § Present address: Eli Lilly & Co., Lilly Corporate Center, DC 4813, Indianapolis, IN 46285. | National Institute on Drug Abuse. # Naval Research Laboratory. ⊥ Present address: PDB, Rutgers, the State University of New Jersey, Piscataway, NJ 08854.

10.1021/jm0305873

amine-induced subjective effects in humans.6 There are, presently, no specific treatment agents available for stimulant abuse, although GBR 12909 is currently being evaluated as a potential pharmacotherapy for that purpose7,8 and GBR 12935 has been considered for use as a treatment agent. High-affinity dopamine uptake inhibitors, such as GBR 12909, selectively block the ability of methamphetamine9 to increase extracellular dopamine. We believe that this effect follows in part from the ability of GBR 12909 to persistently occupy DA transporter binding sites.9 However, a limitation of such agents is that they would not protect against the adverse effects of the amphetamine-like stimulants that are mediated by 5-HT and NE transporters. Therefore, we sought to develop a nonselective transporter ligand that would bind with high affinity to all three biogenic amine transporters. This type of medication would ideally neutralize the neurochemical effects of stimulants such as methamphetamine and not only attenuate the addictive effects of these agents but also block cardiovascular and neurotoxic effects.10 Moreover, we wanted this agent to be amenable to formulation as a long-acting depot medication. One method that has been used to prepare an oil-soluble prodrug is to esterify a polar hydroxyl-containing compound with a medium to long chain alkanoic acid.9

This article not subject to U.S. Copyright. Published 2004 by the American Chemical Society Published on Web 04/14/2004

Indanamine Derivatives

Journal of Medicinal Chemistry, 2004, Vol. 47, No. 10 2625

Scheme 1a

a Reagents and conditions: (a) KOH, EtOH/H O; (b) TFA, 130 °C; (c) NaBH , MeOH; (d) SOCl , 1,4-dioxane; (e) CH NH , toluene, 40 2 4 2 3 2 °C; (f) diethylamine, toluene, 40 °C; (g) 48% HBr, reflux or BBr3, CH2Cl2, 0 °C.

Indatraline ((()-trans-3-(3,4-dichlorophenyl)-N-methyl-1-indanamine, 1) is an uptake inhibitor with high

affinity for DA, 5-HT, and NE transporters.10,11 It blocks the transport of methamphetamine into nerve terminals and therefore blocks the ability of methamphetamine to release these transmitters.10 Since indatraline (1) does not contain a hydroxyl group that could serve as the anchor for the medium to long chain ester functionality, we synthesized a series of methoxyl and phenolic bearing analogues. We previously demonstrated11 that the biological activity of indatraline is less sensitive to substitution on the aromatic ring of the indane than elsewhere in the molecule. Further, a methoxy substituent on the C6 position of the indane has the least negative effect on the binding affinity of indatraline (1).11 Therefore, our synthetic efforts were directed toward compounds with C6-monosubstituted oxygencontaining groups. Chemistry Initial attempts to prepare the needed intermediate, 3-(3,4-dichlorophenyl)-6-methoxylindan-1-one (3), following the procedure of Bogeso12 gave us low yields. We then attempted to prepare 3 from the 1-(3,4dichlorophenyl)-3-(3-methoxylphenyl)-1-propen-3-one (2, Scheme 1). Cyclizing ketone 2 with AlCl3 or BF3‚OEt also gave a low yield of 3. However, 3 was obtained in high yield by heating ketone 2 in neat trifluoroacetic acid at 120 °C in a sealed tube. Following a modification of the published procedure,12 3 was first reduced to 3-(3,4-dichlorophenyl)-6-methoxylindan-1-ol (4) as the

cis diastereomer. The alcohol 4 was converted to the corresponding chloride with a cis/trans ratio of 7:3. Reacting the chloroindan mixtures with methylamine or diethylamine in toluene led to the inversion of the stereocenter to afford the N-methylamine 5 or N,Ndiethylamine 6, respectively, both in cis/trans ratios of 3:7. The cis and trans isomers of 5 and 6 were separated and purified by crystallization to give the racemic cis N-methylamine isomer 7 the cis N,Ndiethylamine isomer 8, the trans N-methylamine isomer 9 and the trans N,N-diethylamine isomer 10. Cleavage of the methoxy group in the trans amine 9 gave the phenol 11, and cleavage of the methoxy group in the trans diethylamine 10 afforded the phenolic compound 12. Optical resolution of 9 was initially accomplished through diastereomeric salt formation with S-(+)- and R-(-)-mandelic acids; however, the method was difficult to replicate. For the reproducible resolution of relatively small quantities, trans isomer 9 was reacted with (R)(-)-1-(1-naphthyl)ethylisocyanate13 to give chromatographically separable diastereomeric ureas (13 and 14, Scheme 2). The ureas 13 and 14 were converted to (-)(1S,3R)-9‚HCl and (+)-(1R,3S)-9‚HCl, respectively, by transamination.14 The optical purity of (-)-(1S,3R)-9 and (+)-(1R,3S)-9 was determined by 500 MHz NMR spectra of their diastereomeric ureas 13 and 14 to show 99.8% ee and 97.6% ee, respectively.15 However, to obtain larger quantities of product for future pharmacological studies in the monkey, we developed an easily replicable chemical resolution of racemate 9 using (-)-(R)- and (+)-(S)-O-acetylmandelic acid (Scheme 3). This gave good yields, and multigram quantities, initially of a levorotatory R-(-)-O-acetylmandelate salt from which the trans-base (+)-(1S,3R)trans-3-(3,4-dichlorophenyl)-6-methoxyl-N-methyl-1-indanamine ((+)-(1S,3R)-9) was obtained. A levorotatory hydrochloride salt was prepared from this (+)-base. The filtrates from the (-)-(R)-O-acetylmandelate salt were basified to give a base enriched with (-)-(1R,3S)-trans3-(3,4-dichlorophenyl)-6-methoxyl-N-methyl-1-indan-

2626

Journal of Medicinal Chemistry, 2004, Vol. 47, No. 10

Yu et al.

Scheme 2a

a

Reagents and conditions: (a) (R)-(-)-1-(1-naphthyl)ethyl isocyanate, C6H6, room temp; (b) aniline, LiCl, 150 °C.

Scheme 3a

a Reagents and conditions: (a) R-(-)-O-acetylmandelic acid, acetone; (b) 20% NH OH; (c) 48% HBr, reflux; (d) (1) NH OH, (2) S-(+)4 4 O-acetylmandelic acid, acetone.

amine ((-)-(1R,3S)-9). Treatment with S-(+)-O-acetylmandelic acid, recrystallization of the dextrorotatory diastereomeric salt, and subsequent basification gave multigram quantities of the desired (-)-(1R,3S)-trans-9 as the free base. It was converted to the dextrorotatory hydrochloride salt. The (-)-(1R,3S)-trans-9 base was also converted to its ditoluoyl-D-tartaric acid salt for an X-ray crystallographic study (Figure 1). The optical purity of both enantiomers of trans amine 9 was found to be >99% ee, determined as described above in the chromatographic resolution of 9. Single-crystal X-ray diffraction analysis of the ditoluoyl-D-tartaric acid salt of (-)-(1R,3S)-trans-9 gave de-

finitive proof of the 1R,3S absolute configuration (Figure 1). The fact that the anion was a D-tartrate salt was used to determine the absolution configuration of the cation. The (-)-(1R,3S)-trans-9 and (+)-(1S,3R)-trans-9 were converted to the phenolic compounds (-)-(1R,3S)trans-3-(3,4-dichlorophenyl)-6-hydroxy-N-methyl-1-indanamine ((-)-(1R,3S)-11) and (+)-(1S,3R)-trans-3-(3,4dichlorophenyl)-6-hydroxy-N-methyl-1-indanamine ((+)(1S,3R)-11), respectively, with HBr. Compounds 7, 8, and 10 were not resolved because of their weak affinity for the norepinephrine transporter (Table 1). We found that in order to prepare a long-chain decanoate ester, it was necessary to first protect the

Indanamine Derivatives

Figure 1. Results of the X-ray analysis on (-)-9 drawn from the experimentally determined coordinates with the thermal parameters at the 20% probability level.

secondary amine with a BOC protecting group and then to react the resulting phenolic compound with 2.8 equiv of decanoic anhydride to form the decanoate ester. Removal of the BOC group under mild conditions (CAN, reflux)16 afforded the decanoate ester 15 (Scheme 4). Ester 15 was stable for at least a month at room temperature. Results and Discussion The binding data shown in Table 1 indicate that all of the compounds have the highest affinity at the 5-HT transporter, followed by the DA and then the NE transporter. 3-(3,4-Dichlorophenyl)-6-methoxy-N, N-diethyl-1-indanamines 8, 10, and 12 bind poorly with the NE transporter. N-Methyl-1-indanamines are generally potent at all three transporters. As reported,12 racemic trans amine 9 is more potent than the racemic cis amine 7. Resolution of the trans amine 9 gave the two enantiomers (+)-(1S,3R)-9 and (-)-(1R,3S)-9. The (-)(1S,3R)-9‚HCl is about twice as potent as the racemate at all of the transporters and considerably more potent than its enantiomer (+)-(1R,3S)-9‚HCl. This trend was also observed for the trans phenolic enantiomers (-)(1R,3S)-11 and (+)-(1S,3R)-11, derived from the methoxy compounds (-)-(1R,3S)-9 and (+)-(1S,3R)-9, respectively. On the basis of the fact that there is a significant difference in the binding affinities of the two enantiomers, we next tested the effects of enantiomers (-)(1S,3R)-9‚HCl, (+)-(1R,3S)-9‚HCl and (-)-(1R,3S)-11, (+)-(1S,3R)-11 on the inhibition of DA, 5-HT, and NE uptake (Table 2). In the methoxy series, the (+)-(1R,3S)9‚HCl enantiomer is better able to inhibit the uptake of neurotransmitters at the DA, 5-HT, and NE transporters, and with the phenolic compounds, the (-)(1R,3S)-11 enantiomer was better. In both cases the enantiomer with the 1R,3S configuration was the more potent ligand. The in vivo activity of (-)-(1R,3S)-11 was assessed using in vivo microdialysis. As shown in Figure 2, iv administration of (-)-(1R,3S)-11 produced significant dose-dependent elevations in extracellular DA (F[3,19] ) 8.05, P < 0.001) and 5-HT (F[3,19] ) 7.11, P < 0.002) in rat nucleus accumbens. The rise in both transmitters was slow in onset and persistent in duration, even with iv administration. Figure 3 shows the effects of pretreatment with (-)-(1R,3S)-11 (0.5 mg/kg, iv) on meth-

Journal of Medicinal Chemistry, 2004, Vol. 47, No. 10 2627

amphetamine-induced (0.5 mg/kg, iv) DA release. As expected, methamphetamine administration to salinepretreated rats produced a rapid and robust increase in extracellular DA. (-)-(1R,3S)-11 significantly blunted the peak effect of methamphetamine on dialysate DA (F[1,20] ) 10.32, P < 0.01). Figure 4 shows the effect of (-)-(1R,3S)-11 on methamphetamine-induced 5-HT release in the same rats. In contrast to the DA data, (-)(1R,3S)-11 pretreatment did not significantly affect the peak effect of methamphetamine. A valid interpretation of the effects of (-)-(1R,3S)-11 pretreatment is complicated by the fact that (-)-(1R,3S)-11 alone increases dialysate transmitter levels. In an attempt to account for this problem, the data in Figure 5 show the effects of (-)-(1R,3S)-11 on methamphetamine-induced transmitter release expressed as a percent of preexisting baseline (i.e., baseline immediately prior to methamphetamine challenge). When the data are expressed in this manner, (-)-(1R,3S)-11 pretreatment significantly reduced the peak effect of methamphetamine on the release of DA (F[1,20] ) 13.13, P 99.4% ee. The (-)-(1S,3R)9‚HCl salt was formed upon treatment of the (+)-(1S,3R)-9 base with HCl in diethyl ether. It was recrystallized from 2-propanol isopropyl ether: mp 225-226 °C; [R]20365 -16.4° (c 0.9, MeOH). Anal. (C17H17Cl2NO‚HCl) C, H, N. The filtrates from the crystallization and recrystallization of the (+)-(1S,3R)-9‚(-)-(R)-O-acetylmandelate salt were combined, evaporated, and converted to the free base (NH4OH, diethyl ether) to give material enriched in (-)-(1R,3S)-9 (21.75 g, 0.0675 mol). (+)-(S)-O-acetylmandelic acid (13.1 g, 0.068 mol) was added to the enriched material in acetone. After standing for 1 h at room temperature, a salt (24.1 g) was obtained that was recrystallized in a manner similar to its diastereomer (see above) to give 20.1 g (73.1%) of the (+)-(S)O-acetylmandelic acid salt of (-)-(1R,3S)-9: mp 177.5-178.5 C; [R]20D +53.4° (c 0.91, MeOH). Anal. (C27H27Cl2NO5) C, H, N. (-)-(1R,3S)-9 was obtained as an oil from the diastereomeric salt in a manner similar to its diastereomeric relative (see above): [R]20365 -13.9° (c 1.68, MeOH). The optical purity of the (-)-(1R,3S)-9 base, determined from the NMR of the urea obtained from reaction with (-)-R-1-(1-naphthyl)ethyl isocyanate, was found to be >99.8% ee. The (-)-(1R,3S)-9 base was converted to its (+)-(1R,3S)‚HCl salt: mp 225-226 °C; [R]20365 +14.85° (c 0.93, MeOH). Anal. (C17H17Cl2NO‚HCl) C, H, N. The (-)-(1R,3S)-9 base was also converted to its di-p-toluoyl-D-(+)tartrate salt by addition of an equimolar amount of acid to the base in MeOH. Recrystallization from MeOH afforded the pure salt: mp 164-166 °C. Anal. (C37H35Cl2NO9‚0.5H2O) C, H, N. (-)-(1R,3S)-trans-3-(3,4-Dichlorophenyl)-6-hydroxy-Nmethyl-1-indanamine ((-)-(1R,3S)-11). A mixture of (-)(1R,3S)-9 (6.3 g, 19.6 mol) and 48% HBr (85 mL) was heated at reflux for 45 min. Most of the HBr was removed in vacuo at 60 °C, the residual oil was dissolved in H2O (150 mL), and the pH was adjusted to 9.5 with concentrated NH4OH. The solid formed after standing at 4 °C overnight was recrystallized from 95% EtOH to give pure (-)-(1R,3S)-11 (2.83 g). An additional amount of (1R,3S)-(-)-11 was obtained by adjustment of the pH of the mother liquor from the above recrystallization to 9.5 with concentrated NH4OH, followed by recrystallization from 95% EtOH (total yield 4.81 g, 79.6%): mp 202-203 °C; [R]20D -19.2° (c 0.9, MeOH); [R]20365 -77.3° (c 0.9, MeOH). Anal. (C16H15Cl2NO) C, H, N. (+)-(1S,3R)-trans-3-(3,4-Dichlorophenyl)-6-hydroxy-Nmethyl-1-indanamine ((+)-(1S,3R)-11). The (+)-(1S,3R)-11 isomer was prepared in 76% yield in a manner similar to (-)(1R,3S)-11 above: mp 200-202 °C; [R]20D +18.3° (c 1.1, MeOH); [R]20365 +76.8° (c 1.1, MeOH). Anal. (C16H15Cl2NO) C, H, N. (()-trans-3-(3,4-Dichlorophenyl)-6-hydroxyl-N,N-diethyl-1-indanamine ((()-12). The compound was prepared from amine (()-10 using the procedure described for (()-11: mp 239 °C; 1H NMR δ 9.74 (s, 1H), 7.61-6.83 (m, 6H), 5.255.22 (m, 1H), 4.59 (t, J ) 8.1 Hz, 1H), 3.32-2.80 (m, 5H), 2.402.29 (m, 1H), 1.31 (t, J ) 6.9 Hz, 3H), 1.22 (t, J ) 6.8 Hz, 3H); CIMS m/z 351 (MH)+. Anal. (C16H15NOCl2) C, H, N, Cl. (()-trans-3-(3,4-Dichlorophenyl)-6-decanoate-N-methyl-1-indanamine ((()-15). To a solution of triethylamine (0.5 mL) in methanol (3 mL) at room temperature was added amine (()-11 (248 mg, 0.81 mmol) in 1 mL of methanol and then ditert-butyl dicarbonate (351 mg, 1.61 mmol). The resulting mixture was stirred at room temperature for 1 h. The solvent

was removed in vacuo, and the product was extracted with CH2Cl2 (2 × 3 mL), washed with water (5 mL), and dried (Na2SO4). The mixture was concentrated to give an oil that was chromatographed to give trans-3-(3,4-dichlorophenyl)-6-hydroxyl-N-methyl-N-(tert-butoxycarbonyl)-1-indanamine (260 mg, 0.64 mmol, 79% yield) as a clear oil: TLC Rf ) 0.40 (20% EtOAc/petroleum ether); 1H NMR δ 7.38-6.71 (m, 6H), 6.015.88 (m, 1H), 4.45-4.35 (m, 1H), 2.79 (s, 3H), 2.79-2.41 (m, 1H), 2.38-2.22 (m, 1H), 1.51 (s, 9H); CIMS m/z 410 (MH)+. To the above amine (260 mg, 0.64 mmol) in 2-propanol (4 mL) was added a solution of NaOH (70 mg, 1.74 mmol) in 1 mL of H2O. Decanoic anhydride (568 mg, 1.74 mmol) was gradually added, and the reaction mixture was stirred for 30 min at room temperature. Solvent was removed in vacuo, and the product was extracted with CH2Cl2 (2 × 3 mL), washed with H2O (5 mL), and dried (Na2SO4). The mixture was concentrated and chromatographed in a mixture of CH2Cl2 and MeOH (9:1) to give trans-3-(3,4-dichlorophenyl)-6-decanoateN-methyl-N-(tert-butoxycarbonyl)-1-indanamine (311 mg, 0.64 mmol, 87% yield) as a clear oil: TLC Rf ) 0.48 (10% EtOAc/ petroleum ether); 1H NMR δ 7.35-6.89 (m, 6H), 6.03-5.78 (m, 1H), 4.48-4.43 (m, 1H), 2.62 (s, 3H), 2.60-2.42 (m, 3H), 2.392.23 (m, 1H), 1.80-1.69 (m, 2H), 1.58 (s, 9H), 1.57-1.21 (m, 12H), 0.89 (t, J ) 5.9 Hz, 3H); CIMS m/z 565 (MH)+. To the above amine (311 mg, 0.64 mmol) in 2.5 mL of acetonitrile was added ammonium cerium nitrate (60 mg, 0.11 mmol). The reaction mixture was stirred at room temperature for 3 h, then was refluxed at 85 °C for another 3 h. The solvent was removed in vacuo and the residue was purified by chromatography to give the deprotected decanoate (()-15 (235 mg, 0.51 mmol, 92% yield) as a clear oil: TLC (50% EtOAc/ petroleum ether) Rf ) 0.42; 1H NMR δ 7.41-6.92 (m, 6H), 4.98-4.88 (m, 1H), 4.68-4.62 (m, 1H), 3.01-2.91 (m, 1H), 2.75 (s, 3H), 2.51 (t, J ) 5.9 Hz, 2H), 2.52-2.37 (m, 1H), 1.75-1.63 (m, 2H), 1.39-1.25 (m, 21H), 0.88 (t, J ) 6.0 Hz, 3H); CIMS m/z 464 (MH)+. Biological Methods. [125I]RTI-55 (0.01 nM, SA ) 2200 Ci/ mmol) was used in the binding assays for the DA (DAT) and 5-HT transporters (SERT).22,23 [3H]nisoxetine (1 nM, SA ) 80 Ci/mmol) was used in the binding assays for the NE transporters (NET).24 All binding assays were performed twice, each in triplicate, using membranes prepared from frozen rat caudate for the DAT and SERT assays and frozen whole rat brain membranes for NET. Briefly, 12 mm × 75 mm polystyrene test tubes were prefilled with 100 µL of drug, 100 µL of radioligand, and 50 µL of a blocker or buffer. Drugs and blockers were made up in 55.2 mM sodium phosphate buffer, pH 7.4 (with the addition of 5 mM KCl and 300 mM NaCl for NET binding), containing 1 mg/mL BSA (BB/BSA). Radioligands were made up in a protease inhibitor cocktail containing BB/BSA, chymostatin (25 µg/mL), leupeptin (25 µg/mL), EDTA (100 µM), and EGTA (100 µM). Membranes added in 750 µL of BB initiated the assay. The samples were incubated for 1824 h at 4 °C (equilibrium) in a final volume of 1 mL. Brandel cell harvesters were used to filter the samples over Whatman GF/B filters, which were presoaked in wash buffer (ice-cold 10 mM Tris-HCl/150 mM NaCl, pH 7.4, containing 2% PEI). The [3H]DA, [3H]5-HT, and [3H]NE uptake assays were run as previously reported.25 All uptake assays were performed twice, each in triplicate, using crude synaptosomes prepared from rat caudate (for DA) or from rat whole brain minus caudate (for 5-HT and NE). Briefly, the assays were initiated by the addition of 100 µL of synaptosomes to 12 mm × 75 mm polystyrene test tubes, which were prefilled with 750 µL of [3H]ligand in a Krebs phosphate buffer (pH 7.4) containing 1 mg/mL ascorbic acid and 50 µM pargyline. [3H]NE assays were performed in the presence of 5 nM 3-(4-iodophenyltropane)2-pyrrolidine carboxamide tartrate to prevent reuptake of [3H]NE into DA nerves. [3H]5-HT uptake assays were performed in the presence of 100 nM nomifensine and 100 nM GBR 12935 to prevent reuptake into NE and DA nerves. The incubations were terminated after 15 min ([3H]DA) or 30 min ([3H]5-HT and [3H]NE) by adding wash buffer (10 mM Tris-HCl/150 mM

Indanamine Derivatives

Journal of Medicinal Chemistry, 2004, Vol. 47, No. 10 2633

NaCl, pH, 7.4) and rapidly filtering over Whatman GF/B filters on a Brandel cell harvester. In vivo microdialysis studies followed published procedures.26 For autoradiography experiments, rats received injections of vehicle (n ) 5 animals), 1 mg/kg (-)-(1R,3S)-11 (n ) 6 animals), or 3 mg/kg (-)-(1R,3S)-11 (n ) 5 animals) and were sacrificed 1 h afterward. By use of published methods, 20 µm coronal sections taken at the caudate level were thaw-mounted onto subbed glass slides, vacuum-desiccated overnight, and stored at -80 °C until assayed. Slide-mounted sections were labeled with [125I]RTI-55 for autoradiographic analysis of transporters. Slides were thawed at room temperature for 5 min, rinsed for 5 min (10 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1 mg/mL bovine serum albumin, 4 °C), and placed into cytomailers containing 10 mL of assay buffer, which consisted of 55.2 mM sodium phosphate buffer (pH 7.4), 1 mg/mL bovine serum albumin, 1 µg/mL chymostatin, 1 µg/mL leupeptin, 100 nM EDTA, 100 nM EGTA, 1 nM [127I]RTI55, 0.01 nM [125I]RTI-55, and appropriate selective transporter blockers. Adjacent sections from each brain were incubated in the absence of blocking drug (total labeling), 100 nM citalopram to select for DA transporter labeling, 1 µM GBR 12909 to select for 5-HT transporter labeling, or 10 µM indatraline to determine nonspecific labeling. After 30 min at 4 °C, slides were rinsed five times for 1 min (10 mM Tris-HCl, pH 7.4, 150 mM NaCl, 1 mg/mL bovine serum albumin, 4 °C) followed by a 10 s rinse in deionized water at 4 °C. Slides were vacuum-desiccated overnight and stored at room temperature until they were analyzed. Autoradiographic distribution of 5-HT and DA transporters was revealed using a Cyclone storage phosphor system (Packard). Slides were placed into photographic film cassettes and apposed to storage phosphor screens of the supersensitive variety. Apposition times were 15 min for DA transporters and 6 h for 5-HT transporters. Transporter levels were quantified by NIH image software, using [125]iodine microscale reference standards (Amersham). DA transporter levels were determined by subtracting nonspecific labeling from labeling in the presence of 100 nM citalopram. 5-HT transporter levels were determined by subtracting nonspecific labeling from labeling in the presence of 1 µM GBR 12909. Results were expressed as the mean SEM. Statistical significance was determine using the Students t-test. Single-Crystal X-ray Diffraction Analysis of (1R,3S)(-)-9‚di-p-toluoyl-D-(+)-tartrate. (C20H17O8)-1, (C17H18Cl2NO)+1, (CH4O), fw ) 740.60 (0.06 mm-1 × 0.20 mm-1 × 0.32 mm-1), orthorhombic space group P212121, a ) 8.0458(19) Å, b )14.451(3) Å, c ) 31.537(5) Å, V ) 3666.8(12) Å3, Z ) 4, dcalc ) 1.34 mg mm-3, λ(Cu KR) ) 1.541 78 Å, µ ) 2.087 mm-1, F(000) ) 1552, T ) 198(2) K, R1 ) 0.0548 for 5650 observed (I > 2σI) reflections and 0.0576 for the full set of 6095 reflections. Data were collected on a Bruker SMART 6K CCD diffractometer using a Rigaku rotating anode source, which was equipped with Gobel mirrors in the incident beam. Corrections were applied for Lorentz, polarization, and absorption effects. The structure was solved and refined with the aid of the programs in the Shelxtl Plus system of programs.27 The fullmatrix least-squares refinement on F2 included atomic coordinates and anisotropic thermal parameters for all non-H atoms. Most of the H atoms were included using a riding model. The fact that the anion was a d-tartrate salt was used to determine the absolution configuration of the cation. The Flack parameter refined to a value of 0.05(2) that confirms the correct absolute structure. Atomic coordinates have been deposited with the Cambridge Crystallographic Database (CCDC 212484), 12 Union Road, Cambridge CB2 1EZ, UK ([email protected]).

crodialysis experiments. The X-ray crystallographic work was supported in part by NIDA, NIH, DHHS, and the Office of Naval Research.

Acknowledgment. The authors (at LMC, NIDDK) thank the National Institute on Drug Abuse (NIDA), NIH, DHHS, for partial financial support of our research program and thank Noel Whittaker (NIDDK) for the mass spectral data and Mario Ayestas, NIDA, NIH, for expert technical assistance in conducting the mi-

Supporting Information Available: Tables 1-6 of X-ray crystal data and structural refinement analysis, atomic coordinates, bond lengths and bond angles, anisotropic displacement parameters, hydrogen coordinates, and hydrogen bond lengths and angles of 1R,3S-(-)-9‚di-p-toluoyl-D-(+)-tartrate. This material is available free of charge via the Internet at http://pubs.acs.org.

References (1) McCoy, C. B.; Inciardi, J. A. Sex, Drugs, and the Continuing Spread of AIDS; Roxbury Publishing Co.: Los Angeles, CA, 1995. (2) McCoy, C. B.; Metsch, L. R.; Inciardi, J. A.; Anwyl, R. S.; Wingerd, J.; Bletzer, K. Sex, Drugs, and the Spread of HIV/AIDS in Belle Glade, Florida. Med. Anthropol. Quart. 1996, 10, 8393. (3) Goodkin, K.; Shapshak, P.; Metsch, L. R.; McCoy, C. B.; Crandall, K. A.; Kumar, M.; Fujimura, R. K.; McCoy, V.; Zhang, B. T.; Reyblat, S.; Xin, K.-Q.; Kumar, A. M. Cocaine Abuse and HIV-1 Infections: Epidemiology and Neuropathogenesis. J. Neuroimmunol. 1998, 83, 88-101 and references therein. (4) Rudnick, G.; Clark, J. From Synapse to Vesicle: The Reuptake and Storage of Biogenic Amine Neurotransmitters. Biochim. Biophys. Acta 1999, 1144, 249. (5) Wise, R. A. Neurobiology of Addiction. Curr. Opin. Neurobiol. 1996, 6, 243-251. (6) Rothman, R. B.; Baumann, M. H.; Dersch, C. M.; Romero, D. V.; Rice, K. C.; Carroll, F. I.; Partilla, J. S. Amphetamine-Type Central Nervous System Stimulants Release Norepinephrine More Potently Than They Release Dopamine and Serotonin. Synapse 2001, 39, 32-41. (7) Zhang, Y. The Identification of GBR 12909 as a Potential Therapeutic Agent for Cocaine Abuse. Med. Chem. Res. 1998, 8, 66-76. (8) Preti, A. Vanoxerine. Curr. Opin. Invest. Drugs 2000, 1, 241251. (9) Baumann, M. H.; Ayestas, M. A.; Sharpe, L. G.; Lewis, D. B.; Rice, K. C.; Rothman, R. B. Persistent Antagonism of Methamphetamine-Induced Dopamine Release in Rats Pretreated with GBR 12909 Decanoate. J. Pharmacol. Exp. Ther. 2002, 301, 1190-1197. (10) Rothman, R. B.; Partilla, J. S.; Bauman, M. H.; Dersch, C. M.; Carroll, F. I.; Rice, K. C. Neurochemical Neutralization of Methamphetamine with High-Affinity Nonselective Inhibitors of Biogenic Amine Transporters: A Pharmacological Strategy for Treating Stimulant Abuse. Synapse 2000, 35, 222-227. (11) Gu, X. H.; Yu, H.; Jacobson, A. E.; Rothman, R. B.; Dersch, C. M.; George, C.; Flippen-Anderson, J. L.; Rice, K. C. Design, Synthesis, and Monoamine Transporter Binding Site Affinities of Methoxy Derivatives of Indatraline. J. Med. Chem. 2000, 43, 4868-4876. (12) Bogeso, K. P.; Christensen, A. V.; Hyttel, J.; Liljefors, T. 3-Phenyl-1-indanamines. Potential Antidepressant Activity and Potent Inhibition of Dopamine, Norepinephrine, and Serotonin Uptake. J. Med. Chem. 1985, 28, 1817-1828. (13) Pirkle, W. H.; Hoekstra, M. S. An Example of Automated Liquid Chromatography. Synthesis of a Broad-Spectrum Resolving Agent and Resolution of 1-(1-Naphthyl)-2,2,2-trifluoroethanol. J. Org. Chem. 1974, 39, 3904-3907. (14) Van Landeghem, H.; De Aquirre, I. Relations between the Magnitude Kinetics and the Thermodynamics of Transformation of Urea. Bull. Soc. Chim. Fr. 1967, 172-178. (15) Rice, K. C.; Brossi, A. Expedient Synthesis of Racemic and Optically Active N-Norreticuline and N-Substituted and 6′Bromo-N-norreticulines. J. Org. Chem. 1980, 45, 592-601. (16) Hwu, J. R.; Jain, M. L.; Tsay, S. C.; Hakimelahi, G. H. Ceric Ammonium Nitrate in the Deprotection of tert-Butoxycarbonyl Group. Tetrahedron Lett. 1996, 37, 2035-2038. (17) Partilla, J. S.; Dersch, C. M.; Yu, H.; Rice, K. C.; Rothman, R. B. Neurochemical Neutralization of Amphetamine-Type Stimulants in Rat Brain by the Indatraline Analog (-)-HY038. Brain Res. Bull. 2001, 53, 821-826. (18) Wyrick, S. D.; Booth, R. G.; Myers, A. M.; Owens, C. E.; Kula, N. S.; Baldessarini, R. J.; McPhail, A. T.; Mailman, R. B. Synthesis and Pharmacological Evaluation of 1-Phenyl-3-amino1,2,3,4-tetrahydronaphthalenes as Ligands for a Novel Receptor with σ-Like Neuromodulatory Activity. J. Med. Chem. 1993, 36, 2542-2551. (19) Southwick, P. L.; Sapper, D. I. 4-Styrylthiazoles. Synthesis and Relationships among Ultraviolet Absorption Spectra. J. Org. Chem. 1954, 19, 1926-1937.

2634

Journal of Medicinal Chemistry, 2004, Vol. 47, No. 10

(20) Greiner, E.; Folk, J. E.; Jacobson, A. E.; Rice, K. C. A Novel and Facile Preparation of Bremazocine Enantiomers through Optically Pure N-Norbremazocines. Bioorg. Med. Chem. 2004, 12, 232-238. (21) Burke, T. R., Jr.; Jacobson, A. E.; Rice, K. C.; Silverton, J. V.; Simonds, W. F.; Streaty, R. A.; Klee, W. A. Probes for Narcotic Receptor Mediated Phenomena. 12. cis-(+)-3-Methylfentanylisothiocyanate, a Potent Site-Directed Acylating Agent for Opioid Recepors. Synthesis, Absolute Configuration, and Receptor Enantioselectivity. J. Med. Chem. 1986, 29, 1087-1093. (22) Matecka, D.; Lewis, D.; Rothman, R. B.; Dersch, C. M.; Wojnicki, F. H. E.; Glowa, J. R.; DeVries, A. C.; Pert, A.; Rice, K. C. Heteroaromatic Analogs of 1-[2-(Diphenylmethoxy)ethyl]- and 1-[2[Bis(4-fluorophenyl)methoxy]ethyl]-4-(3-phenylpropyl)piperazines (GBR 12935 and GBR 12909) as High-Affinity Dopamine Reuptake Inhibitors. J. Med. Chem. 1997, 40, 705-716. (23) Rothman, R. B.; Cadet, J. L.; Akunne, H. C.; Silverthorn, M. L.; Baumann, M. H.; Carroll, F. I.; Goodman, C. B.; Rice, K. C.; de Costa, B. R.; Partilla, J. S.; Wang, J.-B.; Uhl, G. R.; Glowa, J. R.; Dersch, C. M. Studies of the Biogenic Amine Transporters. IV. Demonstration of a Multiplicity of Binding Sites in Rat

Yu et al.

(24)

(25)

(26)

(27)

Caudate Membranes for the Cocaine Analog [125i]RTI-55. J. Pharmacol. Exp. Ther. 1994, 270, 296-309. Rothman, R. B.; Lewis, B.; Dersch, C.; Xu, H.; Radesca, L.; de Costa, B. R.; Rice, K. C.; Kilburn, R. B.; Akunne, H. C.; Pert, A. Identification of a GBR 12935 Homolog, LR 1111, Which Is Over 4,000-Fold Selective for the Dopamine Transporter, Relative to Serotonin and Norepinephrine Transporters. Synapse 1993, 14, 34-39. Rothman, R. B.; Katsnelson, M.; Vu, N.; Partilla, J. S.; Dersch, C. M.; Blough, B. E.; Baumann, M. H. Interaction of the Anorectic Medication, Phendimetrazine, and Its Metabolites with Monoamine Transporters in Rat Brain. Eur. J. Pharmacol. 2002, 47, 51-57. Baumann, M. H.; Phillips, J. M.; Ayestas, M. A.; Ali, S. F.; Rice, K. C.; Rothman, R. B. Preclinical Evaluation of GBR 12909 Decanoate as a Long-Acting Medication for Methamphetamine Dependence. Ann. N. Y. Acad. Sci. 2002, 965, 92-108. Sheldrick, G. M. Shelxtl Plus, version 6.1; Bruker Analytical X-ray Instruments: 2000 Madison, WI.

JM0305873